Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Biotechnol Bioeng ; 109(7): 1844-54, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22252902

RESUMEN

Shear stress is an important physical factor that regulates proliferation, migration, and morphogenesis. In particular, the homeostasis of blood vessels is dependent on shear stress. To mimic this process ex vivo, efforts have been made to seed scaffolds with vascular and other cell types in the presence of growth factors and under pulsatile flow conditions. However, the resulting bioreactors lack information on shear stress and flow distributions within the scaffold. Consequently, it is difficult to interpret the effects of shear stress on cell function. Such knowledge would enable researchers to improve upon cell culture protocols. Recent work has focused on optimizing the microstructural parameters of the scaffold to fine tune the shear stress. In this study, we have adopted a different approach whereby flows are redirected throughout the bioreactor along channels patterned in the porous scaffold to yield shear stress distributions that are optimized for uniformity centered on a target value. A topology optimization algorithm coupled to computational fluid dynamics simulations was devised to this end. The channel topology in the porous scaffold was varied using a combination of genetic algorithm and fuzzy logic. The method is validated by experiments using magnetic resonance imaging readouts of the flow field.


Asunto(s)
Reactores Biológicos , Estrés Mecánico , Andamios del Tejido/química , Algoritmos , Simulación por Computador , Lógica Difusa , Hidrodinámica , Modelos Químicos , Porosidad , Resistencia al Corte
2.
Proc Natl Acad Sci U S A ; 98(22): 12485-90, 2001 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-11606713

RESUMEN

Growth of tumors and metastasis are processes known to require neovascularization. To ascertain the participation of the endogenous angiogenic inhibitor thrombospondin-1 (TSP1) in tumor progression, we generated mammary tumor-prone mice that either lack, or specifically overexpress, TSP1 in the mammary gland. Tumor burden and vasculature were significantly increased in TSP1-deficient animals, and capillaries within the tumor appeared distended and sinusoidal. In contrast, TSP1 overexpressors showed delayed tumor growth or lacked frank tumor development (20% of animals); tumor capillaries showed reduced diameter and were less frequent. Interestingly, absence of TSP1 resulted in increased association of vascular endothelial growth factor (VEGF) with its receptor VEGFR2 and higher levels of active matrix metalloproteinase-9 (MMP9), a molecule previously shown to facilitate both angiogenesis and tumor invasion. In vitro, enzymatic activation of proMMP9 was suppressed by TSP1. Together these results argue for a protective role of endogenous inhibitors of angiogenesis in tumor growth and implicate TSP1 in the in vivo regulation of metalloproteinase-9 activation and VEGF signaling.


Asunto(s)
Factores de Crecimiento Endotelial/metabolismo , Linfocinas/metabolismo , Neoplasias Mamarias Animales/prevención & control , Inhibidores de la Metaloproteinasa de la Matriz , Trombospondina 1/fisiología , Animales , Activación Enzimática , Femenino , Masculino , Ratones , Ratones Transgénicos , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
3.
Mech Dev ; 108(1-2): 161-4, 2001 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11578869

RESUMEN

Mice with targeted mutations in genes required for Notch signal transduction die during embryogenesis, displaying overt signs of hemorrhage due to defects in their vascular development. Surprisingly, directed expression of a constitutively active form of Notch4 within mouse endothelial cells produces a similar vascular embryonic lethality. Moreover, patients with mutations in Notch3 exhibit the cerebral vascular disorder, cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL). These findings underscore the importance of Notch signaling in vascular development; however, they do not identify the specific functional defect. Here, we report that Notch1, Notch3, Notch4, Delta4, Jagged1 and Jagged2 are all expressed in arteries, but are not expressed by veins. These findings identify an aspect of Notch signaling that could contribute to the mechanism by which this pathway modulates vascular morphogenesis.


Asunto(s)
Arterias/embriología , Proteínas de la Membrana/genética , Proteínas de la Membrana/fisiología , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/fisiología , Animales , Arterias/anomalías , Regulación del Desarrollo de la Expresión Génica , Humanos , Inmunohistoquímica , Hibridación in Situ , Ligandos , Ratones , Mutación , Fenotipo , Receptores Notch , Transducción de Señal
4.
Am J Pathol ; 158(4): 1399-410, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11290558

RESUMEN

Regeneration of the endometrium after menstruation requires a rapid and highly organized vascular response. Potential regulators of this process include members of the vascular endothelial growth factor (VEGF) family of proteins and their receptors. Although VEGF expression has been detected in the endometrium, the relationship between VEGF production, receptor activation, and endothelial cell proliferation during the endometrial cycle is poorly understood. To better ascertain the relevance of VEGF family members during postmenstrual repair, we have evaluated ligands, receptors, and activity by receptor phosphorylation in human endometrium throughout the menstrual cycle. We found that VEGF is significantly increased at the onset of menstruation, a result of the additive effects of hypoxia, transforming growth factor-alpha, and interleukin-1beta. Both VEGF receptors, FLT-1 and KDR, followed a similar pattern. However, functional activity of KDR, as determined by phosphorylation studies, revealed activation in the late menstrual and early proliferative phases. The degree of KDR phosphorylation was inversely correlated with the presence of sFLT-1. Endothelial cell proliferation analysis in endometrium showed a peak during the late menstrual and early proliferative phases in concert with the presence of VEGF, VEGF receptor phosphorylation, and decrease of sFLT-1. Together, these results suggest that VEGF receptor activation and the subsequent modulation of sFLT-1 in the late menstrual phase likely contributes to the onset of angiogenesis and endothelial repair in the human endometrium.


Asunto(s)
Endometrio/irrigación sanguínea , Proteínas de la Matriz Extracelular/fisiología , Menstruación/fisiología , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores de Factores de Crecimiento/metabolismo , Adulto , Vasos Sanguíneos/fisiología , Células Cultivadas , Endometrio/citología , Factores de Crecimiento Endotelial/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Proteínas de la Matriz Extracelular/farmacología , Femenino , Humanos , Linfocinas/metabolismo , Fosforilación/efectos de los fármacos , Receptores de Factores de Crecimiento Endotelial Vascular , Solubilidad , Factores de Tiempo , Factor A de Crecimiento Endotelial Vascular , Receptor 1 de Factores de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
5.
J Biol Chem ; 276(16): 13372-8, 2001 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-11278559

RESUMEN

Mature human aorta contains a 70-kDa versican fragment, which reacts with a neoepitope antiserum to the C-terminal peptide sequence DPEAAE. This protein therefore appears to represent the G1 domain of versican V1 (G1-DPEAAE(441)), which has been generated in vivo by proteolytic cleavage at the Glu(441)-Ala(442) bond, within the sequence DPEAAE(441)-A(442)RRGQ. Because the equivalent aggrecan product (G1-NITEGE(341)) and brevican product (G1-EAVESE(395)) are generated by ADAMTS-mediated cleavage of the respective proteoglycans, we tested the capacity of recombinant ADAMTS-1 and ADAMTS-4 to cleave versican at Glu(441)-Ala(442). Both enzymes cleaved a recombinant versican substrate and native human versican at the Glu(441)-Ala(442) bond and the mature form of ADAMTS-4 was detected by Western analysis of extracts of aortic intima. We conclude that versican V1 proteolysis in vivo can be catalyzed by one or more members of the ADAMTS family of metalloproteinases.


Asunto(s)
Alanina , Aorta Abdominal/metabolismo , Proteoglicanos Tipo Condroitín Sulfato/química , Proteoglicanos Tipo Condroitín Sulfato/metabolismo , Desintegrinas/metabolismo , Ácido Glutámico , Metaloendopeptidasas/metabolismo , Músculo Liso Vascular/metabolismo , Proteínas ADAM , Proteína ADAMTS1 , Proteína ADAMTS4 , Células Cultivadas , Humanos , Lectinas Tipo C , Datos de Secuencia Molecular , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Procolágeno N-Endopeptidasa , Proteoglicanos/metabolismo , Proteínas Recombinantes/metabolismo , Especificidad por Sustrato , Túnica Íntima/metabolismo , Versicanos
6.
Hum Reprod ; 15 Suppl 3: 39-47, 2000 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-11041220

RESUMEN

Progestin-only contraceptives are associated with breakthrough bleeding in up to 50% of users. The causes of blood vessel rupture are not well understood. Here we report that both normal and Norplant-exposed endothelium express progesterone receptor. Experiments performed in vitro on endothelial cells isolated from human endometrium revealed that longterm progesterone exposure leads to suppression of endothelial cell proliferation, inhibition of migration and alteration in the profile of extracellular matrix proteins secreted by human endometrial endothelial cells. In addition, we detected increased levels of matrix metalloproteinase-9 in endothelial cultures treated with progesterone. The effect of progesterone on the cell cycle, along with the increased amounts of matrix-degrading enzymes, could account for breakdown of basement membrane components, vascular fragility and consequent vessel rupture leading to breakthrough endometrial bleeding.


Asunto(s)
Endometrio/irrigación sanguínea , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/fisiología , Progesterona/farmacología , Receptores de Progesterona/efectos de los fármacos , Receptores de Progesterona/fisiología , Adolescente , Adulto , Membrana Basal/efectos de los fármacos , Endotelio Vascular/enzimología , Femenino , Humanos , Levonorgestrel/farmacología , Metaloproteinasa 9 de la Matriz/metabolismo , Congéneres de la Progesterona/farmacología , Promegestona/farmacología , ARN Mensajero/análisis , Receptores de Progesterona/genética , Hemorragia Uterina/inducido químicamente , Hemorragia Uterina/fisiopatología
7.
Mol Biol Cell ; 11(9): 2885-900, 2000 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10982388

RESUMEN

Thrombospondin-1 (TSP1) can inhibit angiogenesis by interacting with endothelial cell CD36 or proteoglycan receptors. We have now identified alpha3beta1 integrin as an additional receptor for TSP1 that modulates angiogenesis and the in vitro behavior of endothelial cells. Recognition of TSP1 and an alpha3beta1 integrin-binding peptide from TSP1 by normal endothelial cells is induced after loss of cell-cell contact or ligation of CD98. Although confluent endothelial cells do not spread on a TSP1 substrate, alpha3beta1 integrin mediates efficient spreading on TSP1 substrates of endothelial cells deprived of cell-cell contact or vascular endothelial cadherin signaling. Activation of this integrin is independent of proliferation, but ligation of the alpha3beta1 integrin modulates endothelial cell proliferation. In solution, both intact TSP1 and the alpha3beta1 integrin-binding peptide from TSP1 inhibit proliferation of sparse endothelial cell cultures independent of their CD36 expression. However, TSP1 or the same peptide immobilized on the substratum promotes their proliferation. The TSP1 peptide, when added in solution, specifically inhibits endothelial cell migration and inhibits angiogenesis in the chick chorioallantoic membrane, whereas a fragment of TSP1 containing this sequence stimulates angiogenesis. Therefore, recognition of immobilized TSP1 by alpha3beta1 integrin may stimulate endothelial cell proliferation and angiogenesis. Peptides that inhibit this interaction are a novel class of angiogenesis inhibitors.


Asunto(s)
Comunicación Celular/fisiología , Endotelio Vascular/fisiología , Integrinas/fisiología , Trombospondina 1/fisiología , Alantoides/irrigación sanguínea , Alantoides/fisiología , Animales , Aorta , Antígenos CD36/fisiología , Cadherinas/fisiología , Bovinos , División Celular , Células Cultivadas , Embrión de Pollo , Corion/irrigación sanguínea , Corion/fisiología , Endotelio Vascular/citología , Fibronectinas/farmacología , Fibronectinas/fisiología , Integrina alfa3beta1 , Neovascularización Fisiológica/efectos de los fármacos , Neovascularización Fisiológica/fisiología , Transducción de Señal , Trombospondina 1/farmacología
8.
J Biol Chem ; 275(43): 33471-9, 2000 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-10944521

RESUMEN

METH-1/ADAMTS1 is a member of a newly described family of genes that contain metalloprotease, disintegrin, and thrombospondin-like motifs. We have recently shown that METH-1 protein is a potent inhibitor of angiogenesis. Here, we demonstrate that secreted human pro-METH-1 is processed in two consecutive steps to release both p87 and p65 active forms. The p87 form lacks the N-terminal prodomain and p65 results from an additional processing event in the C-terminal end. Generation of p87 was blocked with specific inhibitors of furin, and incubation of pro-METH-1 with purified furin released the p87 fragment but not p65. Generation of p65 required preformation of p87 and was suppressed by inhibitors of matrix metalloproteases. We demonstrate that matrix metalloproteases 2, 8, and 15 were able to release p65 when p87 was used as substrate. This second processing step removes two thrombospondin repeats from the carboxyl-terminal end of p87-METH-1 and alters the affinity of the protein to heparin and endothelial cultures. Furthermore, this deletion was associated with a reduced activity upon suppression of endothelial cell proliferation. We hypothesize that METH-1 processing is relevant for the modulation of the anti-angiogenic properties displayed by the protein.


Asunto(s)
Desintegrinas , Metaloendopeptidasas/metabolismo , Proteínas ADAM , Proteína ADAMTS1 , Animales , Sitios de Unión , Bovinos , Células Cultivadas , Endotelio Vascular/metabolismo , Furina , Humanos , Metaloendopeptidasas/fisiología , Subtilisinas/fisiología , Zinc/metabolismo
9.
Microvasc Res ; 60(2): 112-20, 2000 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10964585

RESUMEN

All-trans-retinoic acid (RA) and other retinoids modulate cell growth and differentiation, generally favoring terminal cell differentiation and inhibiting carcinogenesis. Retinoids are also reported to inhibit angiogenesis and endothelial cell migration, actions that are also anti-carcinogenic. Vascular permeability factor/vascular endothelial growth factor (VPF/VEGF) is a multifunctional cytokine secreted by many tumors. It renders microvessels hyperpermeable to plasma and stimulates endothelial cell migration and division. To investigate further the mechanisms by which RA inhibits angiogenesis, we evaluated the effects of RA on VPF/VEGF-induced angiogenesis and microvascular permeability. RA selectively inhibited the angiogenic response induced by VPF/VEGF, but not that induced by fibroblast growth factor-2 (FGF-2), in the CAM assay. RA and two of its isomers also inhibited the vascular permeabilizing effect of VPF/VEGF but not that induced by histamine. The vascular permeabilization induced by VPF/VEGF and blocked by RA takes place within 1-15 min, too short a time frame for RA to act by modulating transcription through classic retinoid receptors. RA also inhibited VPF/VEGF-induced phosphorylation of PLC-gamma and synthesis of cGMP but actually increased VPF/VEGF binding to cultured endothelial cells. Taken together, these findings indicate that RA selectively blocks VPF/VEGF-induced microvascular permeability and angiogenesis and also identify VPF/VEGF as a major target of RA action. The selectivity of RA's action suggests that other, RA-independent pathways must exist for the angiogenesis induced by FGF-2 and the vascular permeabilizing effect of histamine.


Asunto(s)
Antineoplásicos/farmacología , Permeabilidad Capilar/efectos de los fármacos , Factores de Crecimiento Endotelial/farmacología , Linfocinas/farmacología , Neovascularización Fisiológica/efectos de los fármacos , Tretinoina/farmacología , Animales , Bioensayo , Antagonismo de Drogas , Cobayas , Masculino , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
10.
Dev Dyn ; 218(3): 507-24, 2000 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-10878616

RESUMEN

Vascular endothelial growth factors (VEGFs) are endothelial cell-specific mitogens with potent angiogenic and vascular permeability-inducing properties. VEGF, VEGF-C, and VEGFRs -1, -2, and -3 were found to be expressed in post-pubertal (virgin) rodent mammary glands. VEGF was increased during pregnancy (5-fold) and lactation (15-19-fold). VEGF-C was moderately increased during pregnancy and lactation (2- and 3-fold respectively). VEGF levels were reduced by approximately 75% in cleared mouse mammary glands devoid of epithelial components, demonstrating that although the epithelial component is the major source of VEGF, approximately 25% is derived from stroma. This was confirmed by the findings (a) that VEGF transcripts were expressed predominantly in ductal and alveolar epithelial cells, and (b) that VEGF protein was localized to ductal epithelial cells as well as to the stromal compartment including vascular structures. VEGF was detected in human milk. Finally, transcripts for VEGFRs -2 and -3 were increased 2-3-fold during pregnancy, VEGFRs -1, -2 and -3 were increased 2-4-fold during lactation, and VEGFRs -2 and -3 were decreased by 20-50% during involution. These results point to a causal role for the VEGF ligand-receptor pairs in pregnancy-associated angiogenesis in the mammary gland, and suggest that they may also regulate vascular permeability during lactation.


Asunto(s)
Factores de Crecimiento Endotelial/metabolismo , Regulación de la Expresión Génica/fisiología , Lactancia/fisiología , Linfocinas/metabolismo , Glándulas Mamarias Animales/fisiología , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores de Factores de Crecimiento/metabolismo , Adulto , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Northern Blotting , Capilares/crecimiento & desarrollo , Capilares/fisiología , Línea Celular , Factores de Crecimiento Endotelial/genética , Factores de Crecimiento Endotelial/inmunología , Femenino , Humanos , Inmunohistoquímica , Hibridación in Situ , Lactancia/genética , Linfocinas/genética , Linfocinas/inmunología , Glándulas Mamarias Animales/anatomía & histología , Glándulas Mamarias Animales/irrigación sanguínea , Ratones , Leche Humana/química , Datos de Secuencia Molecular , Neovascularización Fisiológica , Embarazo , ARN/aislamiento & purificación , ARN/metabolismo , Ratas , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/inmunología , Receptores de Factores de Crecimiento/genética , Receptores de Factores de Crecimiento/inmunología , Receptores de Factores de Crecimiento Endotelial Vascular , Alineación de Secuencia , Factor A de Crecimiento Endotelial Vascular , Factor C de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular , Destete
11.
Mol Cell Biol ; 20(5): 1723-32, 2000 03.
Artículo en Inglés | MEDLINE | ID: mdl-10669749

RESUMEN

Accumulating evidence suggests that phosphatases play an important role in regulating a variety of signal transduction pathways that have a bearing on cancer. The kinase-associated phosphatase (KAP) is a human dual-specificity protein phosphatase that was identified as a Cdc2- or Cdk2-interacting protein by a yeast two-hybrid screening, yet the biological significance of these interactions remains elusive. We have identified the KAP gene as an overexpressed gene in breast and prostate cancer by using a phosphatase domain-specific differential-display PCR strategy. Here we report that breast and prostate malignancies are associated with high levels of KAP expression. The sublocalization of KAP is variable. In normal cells, KAP is primarily found in the perinuclear region, but in tumor cells, a significant portion of KAP is found in the cytoplasm. Blocking KAP expression by antisense KAP in a tetracycline-regulatable system results in a reduced population of S-phase cells and reduced Cdk2 kinase activity. Furthermore, lowering KAP expression led to inhibition of the transformed phenotype, with reduced anchorage-independent growth and tumorigenic potential in athymic nude mice. These findings suggest that therapeutic intervention might be aimed at repression of KAP gene overexpression in human breast and prostate cancer.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Proteínas de Ciclo Celular , Transformación Celular Neoplásica/genética , Oligonucleótidos Antisentido/genética , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Proteínas Tirosina Fosfatasas/genética , Animales , Diferenciación Celular/genética , Proteínas Inhibidoras de las Quinasas Dependientes de la Ciclina , Fosfatasas de Especificidad Dual , Femenino , Humanos , Masculino , Ratones , Oligonucleótidos Antisentido/farmacología , Proteínas Tirosina Fosfatasas/antagonistas & inhibidores , Células Tumorales Cultivadas
12.
Cancer Metastasis Rev ; 19(1-2): 159-65, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-11191055

RESUMEN

Angiogenesis has been acknowledged as an important requirement for growth and metastasis of tumors. Complete or partial suppression of vascular growth by a number of different strategies has been consistently associated with suppression of tumor expansion and even reduction of tumor burden. Consequently, identification of the molecular pathways of the angiogenic response has been a major focus of interest in academia and industry. The development of tumor-specific anti-angiogenic therapy was also catalyzed by the finding that inhibitors of angiogenesis appeared immune to the development of drug resistance by the tumor cells, a major restrain in current chemotherapy. Although the full identification of players and their cross-talk is still at its infancy, it appears that partial blockade of one of the steps in the angiogenesis cascade, is sufficient to affect capillary morphogenesis. Thus, suppression of specific integrin pathways or vascular endothelial growth factor signaling have been shown effective in the suppression of tumor-mediated angiogenesis and have led to subsequent initiation of clinical trials. In addition to the generation of antibodies or chemical mimetics to interfere with particular steps during vascular organization, several endogenous (or physiological) molecules have also been identified. The list of endogenous modulators of angiogenesis is growing and can offer additional and important tool for the generation of therapies to restrain tumor vascularization. This review will focus one group of such molecules which include the thrombospondins and metallospondins, two families of proteins linked by the presence of a conserved anti-angiogenic functional domain.


Asunto(s)
Neoplasias/irrigación sanguínea , Neoplasias/patología , Neovascularización Patológica/fisiopatología , Neovascularización Fisiológica/fisiología , Trombospondina 1/fisiología , Proteínas ADAM , Proteína ADAMTS1 , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Desintegrinas/metabolismo , Humanos , Metaloendopeptidasas/metabolismo , Ratones , Metástasis de la Neoplasia , Neoplasias/fisiopatología , Neovascularización Patológica/prevención & control
13.
Circulation ; 100(13): 1423-31, 1999 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-10500044

RESUMEN

BACKGROUND: Suppression of tumor growth by thrombospondin-1 (TSP-1) has been associated with its ability to inhibit neovascularization. The antiangiogenic activity of TSP-1, as defined by cornea pocket assays, was previously mapped to the amino-terminal portion of the protein within the procollagen region and the type 1 repeats. METHODS AND RESULTS: We evaluated the specificity and efficacy of different regions of TSP-1 using recombinant fragments of the protein on chorioallantoic membrane (CAM) angiogenesis and endothelial cell proliferation assays. In both assays, fragments containing the second and third type 1 repeats but not the procollagen region inhibited angiogenesis and endothelial cell proliferation. To further define the sequences responsible for the angiostatic effect of TSP-1, we used synthetic peptides. The CAM assay defined 2 sequences that independently suppressed angiogenesis. The amino-terminal end of the type 1 repeats showed higher potency for inhibiting angiogenesis driven by basic fibroblast growth factor (FGF-2), whereas the second region equally blocked angiogenesis driven by either FGF-2 or vascular endothelial growth factor (VEGF). Modifications of the active peptides revealed the specific amino acids required for the inhibitory response. One sequence included the conserved tryptophan residues in the amino-terminal end of the second and third type 1 repeats, and the other involved the amino acids that follow the CSVTCG sequence in the carboxy-terminus of these repeats. Both inhibition in the CAM assay and inhibition of breast tumor xenograft growth in nude mice were independent of the TGF-beta-activating sequence located in the second type 1 repeat. CONCLUSIONS: These results indicate that the type 1 repeats of TSP-1 contain 2 subdomains that may independently inhibit neovascularization. They also identify 2 independent pathways by which TSP-1 can block FGF-2 and VEGF angiogenic signals on endothelial cells.


Asunto(s)
Neovascularización Patológica/prevención & control , Trombospondina 1/genética , Trombospondina 1/farmacología , Secuencia de Aminoácidos/genética , Animales , División Celular/efectos de los fármacos , Embrión de Pollo , Factores de Crecimiento Endotelial/antagonistas & inhibidores , Factores de Crecimiento Endotelial/fisiología , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Membranas Extraembrionarias/irrigación sanguínea , Membranas Extraembrionarias/efectos de los fármacos , Femenino , Factor 2 de Crecimiento de Fibroblastos/antagonistas & inhibidores , Factor 2 de Crecimiento de Fibroblastos/fisiología , Linfocinas/antagonistas & inhibidores , Linfocinas/fisiología , Neoplasias Mamarias Animales/patología , Ratones , Ratones Desnudos , Datos de Secuencia Molecular , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/farmacología , Proteínas Recombinantes , Secuencias Repetitivas de Aminoácido , Transducción de Señal/efectos de los fármacos , Trasplante Heterólogo , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
14.
J Biol Chem ; 274(33): 23349-57, 1999 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-10438512

RESUMEN

We have studied two related proteins that contain a repeated amino acid motif homologous to the anti-angiogenic type 1 repeats of thrombospondin-1 (TSP1). Complete sequence analysis revealed no other similarities with TSP1, but identified unique signal sequences, as well as metalloprotease and disintegrin-like domains in the NH(2) termini. We named these proteins METH-1 and METH-2 due to the novel combination of metalloprotease and thrombospondin domains. Overall amino acid sequence identity between METH-1 and METH-2 is 51. 7%, yet transcript distribution revealed non-overlapping patterns of expression in tissues and cultured cell lines. To characterize these proteins functionally, we isolated full-length cDNAs, produced recombinant protein, and generated antisera to the recombinant proteins. Both METH-1 and METH-2 represent single copy genes, which encode secreted and proteolytically processed proteins. METH proteins suppressed fibroblast growth factor-2-induced vascularization in the cornea pocket assay and inhibited vascular endothelial growth factor-induced angiogenesis in the chorioallantoic membrane assay. Suppression of vessel growth in both assays was considerably greater than that mediated by either thrombospondin-1 or endostatin on a molar basis. Consistent with an endothelial specific response, METH-1 and METH-2 were shown to inhibit endothelial cell proliferation, but not fibroblast or smooth muscle growth. We propose that METH-1 and METH-2 represent a new family of proteins with metalloprotease, disintegrin, and thrombospondin domains. The distinct distribution of each gene product suggests that each has evolved distinct regulatory mechanisms that potentially allow for fine control of activity during distinct physiological and pathological states.


Asunto(s)
Desintegrinas/genética , Metaloendopeptidasas/genética , Metaloendopeptidasas/fisiología , Neovascularización Fisiológica , Proteínas ADAM , Proteínas ADAMTS , Proteína ADAMTS1 , Secuencia de Aminoácidos , División Celular/fisiología , Células Cultivadas , Clonación Molecular , ADN Complementario , Desintegrinas/química , Humanos , Metaloendopeptidasas/química , Datos de Secuencia Molecular , Sistemas de Lectura Abierta , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Homología de Secuencia de Aminoácido
15.
Microcirculation ; 6(2): 127-40, 1999 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10466115

RESUMEN

OBJECTIVE: To develop a reliable method for the isolation and longterm culture of microvessel endothelial cells from human endometrium and to evaluate their response to angiogenic growth factors and steroid hormones in comparison to endothelial cells derived from other organs. METHODS: Endometrial tissue from hysterectomy specimens were digested sequentially with collagenase and trypsin, cultured for 24 h, then selected by adhesion to anti-CD-34 coated magnetic beads. Alternatively, anti-CD-34-coated beads could also be substituted by Ulex europaeus agglutinin-1, anti-PECAM, or anti-E-selectin-coated beads. Characterization of the isolated cultures included expression of endothelial cell markers, regulation of E-selectin in response to TNF-alpha, proliferative response to angiogenic growth factors, and expression of progesterone and estrogen receptors. We also analyzed the relative binding affinity of VEGF on endometrial endothelial cells in comparison to other endothelial cell types. RESULTS: Selection on anti-CD-34-coated beads eliminated contaminating cells and resulted in a homogeneous population of human endometrial endothelial cells (HEEC), as assessed by expression of PECAM, von Willebrand's factor, and uptake of acetylated-LDL. HEEC also upregulated E-selectin in response to TNF-alpha in a manner similar to that seen for other endothelial cell types. Expression of progesterone and estrogen receptor was revealed by immunocytochemistry and RT-PCR consistently until passage 5. Endometrial endothelial cells were more responsive to growth stimulation by VEGF than were dermal endothelial cells isolated under similar conditions. Further characterization indicated that VEGF bound more avidly to HEEC than to other endothelial cell types. CONCLUSIONS: Human endometrial endothelial cells were isolated to homogeneity by a two-part protocol and successfully passaged under culture conditions similar to those used for other endothelial cell types. The HEEC were very responsive to VEGF growth-stimulation likely due to elevated affinity, or increased levels of, KDR and FLT-1 on the cell surface. These results indicate that HEEC are capable of maintaining a mature phenotype in culture and might provide a model for understanding the response of these cells to the recurrent cycles of proliferation imposed on the endometrium during menstruation.


Asunto(s)
Endometrio/irrigación sanguínea , Factores de Crecimiento Endotelial/farmacología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Linfocinas/farmacología , Receptores de Estrógenos/metabolismo , Receptores de Progesterona/metabolismo , Técnicas de Cultivo de Célula/métodos , División Celular/efectos de los fármacos , Separación Celular/métodos , Factores de Crecimiento Endotelial/metabolismo , Endotelio Vascular/citología , Estradiol/farmacología , Femenino , Factor 2 de Crecimiento de Fibroblastos/farmacología , Humanos , Separación Inmunomagnética , Linfocinas/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Progesterona/farmacología , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
16.
Development ; 126(16): 3597-605, 1999 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-10409505

RESUMEN

Erythropoietin is an essential growth factor that promotes survival, proliferation, and differentiation of mammalian erythroid progenitor cells. Erythropoietin(-/-) and erythropoietin receptor(-/-) mouse embryos die around embryonic day 13.5 due, in part, to failure of erythropoiesis in the fetal liver. In this study, we demonstrated a novel role of erythropoietin and erythropoietin receptor in cardiac development in vivo. We found that erythropoietin receptor is expressed in the developing murine heart in a temporal and cell type-specific manner: it is initially detected by embryonic day 10.5 and persists until day 14.5. Both erythropoietin(-/-) and erythropoietin receptor(-/-) embryos suffered from ventricular hypoplasia at day 12-13 of gestation. This defect appears to be independent from the general state of hypoxia and is likely due to a reduction in the number of proliferating cardiac myocytes in the ventricular myocardium. Cell proliferation assays revealed that erythropoietin acts as a mitogen in cells isolated from erythropoietin(-/-) mice, while it has no effect in hearts from erythropoietin receptor(-/-) animals. Erythropoietin(-/-) and erythropoietin receptor(-/-) embryos also suffered from epicardium detachment and abnormalities in the vascular network. Finally, through a series of chimeric analysis, we provided evidence that erythropoietin acts in a manner which is non-cell-autonomous. Our results elucidate a novel role of erythropoietin in cardiac morphogenesis and suggest a combination of anemia and cardiac failure as the cause of embryonic lethality in the erythropoietin(-/-) and erythropoietin receptor(-/-) animals.


Asunto(s)
Eritropoyetina/fisiología , Regulación del Desarrollo de la Expresión Génica , Cardiopatías Congénitas/embriología , Corazón/embriología , Morfogénesis , Receptores de Eritropoyetina/fisiología , Animales , Diferenciación Celular , División Celular , Células Cultivadas , Quimera , Eritropoyetina/deficiencia , Eritropoyetina/genética , Cardiopatías Congénitas/genética , Ratones , Ratones Noqueados , Miocardio/citología , Receptores de Eritropoyetina/deficiencia , Receptores de Eritropoyetina/genética , Células Madre/citología , Células Madre/fisiología
17.
Clin Cancer Res ; 5(5): 1041-56, 1999 May.
Artículo en Inglés | MEDLINE | ID: mdl-10353737

RESUMEN

The generation of vascular stroma is essential for solid tumor growth and involves stimulatory and inhibiting factors as well as stromal components that regulate functions such as cellular adhesion, migration, and gene expression. In an effort to obtain a more integrated understanding of vascular stroma formation in breast carcinoma, we examined expression of the angiogenic factor vascular permeability factor (VPF)/vascular endothelial growth factor (VEGF); the VPF/VEGF receptors flt-1 and KDR; thrombospondin-1, which has been reported to inhibit angiogenesis; and the stromal components collagen type I, total fibronectin, ED-A+ fibronectin, versican, and decorin by mRNA in situ hybridization on frozen sections of 113 blocks of breast tissue from 68 patients including 28 sections of breast tissue without malignancy, 18 with in situ carcinomas, 56 with invasive carcinomas, and 8 with metastatic carcinomas. A characteristic expression profile emerged that was remarkably similar in invasive carcinoma, carcinoma in situ, and metastatic carcinoma, with the following characteristics: strong tumor cell expression of VPF/VEGF; strong endothelial cell expression of VPF/VEGF receptors; strong expression of thrombospondin-1 by stromal cells and occasionally by tumor cells; and strong stromal cell expression of collagen type I, total fibronectin, ED-A+ fibronectin, versican, and decorin. The formation of vascular stroma preceded invasion, raising the possibility that tumor cells invade not into normal breast stroma but rather into a richly vascular stroma that they have induced. Similarly, tumor cells at sites of metastasis appear to induce the vascular stroma in which they grow. We conclude that a distinct pattern of mRNA expression characterizes the generation of vascular stroma in breast cancer and that the formation of vascular stroma may play a role not only in growth of the primary tumor but also in invasion and metastasis.


Asunto(s)
Neoplasias de la Mama/irrigación sanguínea , Carcinoma in Situ/irrigación sanguínea , Carcinoma/irrigación sanguínea , Neovascularización Patológica , Adenocarcinoma Mucinoso/irrigación sanguínea , Adenocarcinoma Mucinoso/química , Adenocarcinoma Mucinoso/patología , Biomarcadores , Biopsia , Mama/irrigación sanguínea , Mama/química , Neoplasias de la Mama/química , Neoplasias de la Mama/patología , Carcinoma/química , Carcinoma/patología , Carcinoma in Situ/química , Carcinoma Ductal de Mama/irrigación sanguínea , Carcinoma Ductal de Mama/química , Carcinoma Ductal de Mama/patología , Carcinoma Intraductal no Infiltrante/irrigación sanguínea , Carcinoma Intraductal no Infiltrante/química , Carcinoma Intraductal no Infiltrante/patología , Carcinoma Lobular/irrigación sanguínea , Carcinoma Lobular/química , Carcinoma Lobular/patología , Proteoglicanos Tipo Condroitín Sulfato/análisis , Colágeno/análisis , Decorina , Factores de Crecimiento Endotelial/análisis , Endotelio Vascular/química , Células Epiteliales/química , Proteínas de la Matriz Extracelular , Femenino , Enfermedad Fibroquística de la Mama/metabolismo , Enfermedad Fibroquística de la Mama/patología , Fibronectinas/análisis , Secciones por Congelación , Humanos , Hibridación in Situ , Lectinas Tipo C , Metástasis Linfática , Linfocinas/análisis , Invasividad Neoplásica , Metástasis de la Neoplasia , Proteínas de Neoplasias/análisis , Isoformas de Proteínas/análisis , Proteoglicanos/análisis , Proteínas Proto-Oncogénicas/análisis , ARN Mensajero/análisis , ARN Neoplásico/análisis , Proteínas Tirosina Quinasas Receptoras/análisis , Receptores de Factores de Crecimiento/análisis , Receptores de Factores de Crecimiento Endotelial Vascular , Células del Estroma/patología
18.
J Biol Chem ; 274(4): 2185-92, 1999 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-9890981

RESUMEN

The use of steroid hormones in postmenopausal replacement therapy has been associated with prevention of cardiovascular disease. Although the contribution of estradiol to endothelial cell function has been addressed, little information is available on the effect of progestins on this cell type. Here, we provide direct evidence for the presence of functional nuclear progesterone receptor in endothelial cells and demonstrate that physiological levels of progesterone inhibit proliferation through a nuclear receptor-mediated mechanism. The effects of progesterone were blocked by pretreatment with a progesterone receptor antagonist, and progesterone receptor-deficient endothelial cells failed to respond to the hormone. We evaluated the effect of progesterone by analysis of aorta re-endothelialization experiments in wild-type and progesterone receptor knockout mice. The rate of re-endothelialization was significantly decreased in wild-type mice when in the presence of progesterone, whereas there was no difference between control and progesterone-treated progesterone receptor knockout mice. FACS analysis showed that progestins arrest endothelial cell cycle in G1. The lag in cell cycle progression involved reduction in cyclin-dependent kinase activity, as shown by down-regulation in retinoblastoma protein phosphorylation. In addition, treatment of endothelial cells with progestins altered the expression of cyclin E and A in accordance with G1 arrest. These results have important implications to our current knowledge of the effect of steroids on endothelial cell function and to the overall contribution of progesterone to vascular repair.


Asunto(s)
División Celular/efectos de los fármacos , Endotelio/efectos de los fármacos , Progesterona/farmacología , Animales , Ciclo Celular/efectos de los fármacos , Células Cultivadas , Endotelio/citología , Femenino , Humanos , Ratones , Ratones Noqueados
19.
Cancer Res ; 59(1): 189-97, 1999 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-9892206

RESUMEN

Endostatin is a Mr 20,000 COOH-terminal fragment of collagen XVIII that inhibits the growth of several primary tumors. We report here the cloning and expression of mouse endostatin in both prokaryotic and eukaryotic expression systems. Soluble recombinant protein expressed in yeast (15-20 mg/L) inhibited the proliferation and migration of endothelial cells in response to stimulation by basic fibroblast growth factor. A rabbit polyclonal antibody was raised that showed positive immunoreactivity to the recombinant protein expressed from both systems. Importantly, the biological activity of the mouse recombinant protein could be neutralized by this antiserum in both endothelial proliferation and chorioallantoic membrane assays. Systemic administration of endostatin at 10 mg/kg suppressed the growth of renal cell cancer in a nude mouse model. The inhibition of tumor growth with soluble yeast-produced protein was comparable to that obtained with non-refolded precipitated protein expressed from bacteria. In addition, two closely related COOH-terminal deletion mutants of endostatin were also tested and showed strikingly differing activity. Collectively, these findings demonstrate the expression of a biologically active form of mouse endostatin in yeast, define a role for the molecule in inhibiting endothelial cell migration, extend its antitumor effects to renal cell carcinoma, and provide a formal proof (via the neutralizing antiserum experiments and the mutant data) that endostatin (and not a possible contaminant) acts as an antiangiogenic agent. Finally, the high level expression of mouse endostatin in yeast serves as an endotoxin free, soluble source of protein for fundamental studies on the mechanisms of tumor growth suppression by angiogenesis inhibitors.


Asunto(s)
Antineoplásicos/uso terapéutico , Carcinoma de Células Renales/tratamiento farmacológico , Colágeno/uso terapéutico , Neoplasias Renales/tratamiento farmacológico , Neoplasias Experimentales/tratamiento farmacológico , Fragmentos de Péptidos/uso terapéutico , Animales , Antineoplásicos/farmacología , Carcinoma de Células Renales/patología , Bovinos , Colágeno/biosíntesis , Colágeno/genética , Colágeno/farmacología , Colágeno Tipo XVIII , Endostatinas , Neoplasias Renales/patología , Ratones , Mutación , Neoplasias Experimentales/patología , Fragmentos de Péptidos/biosíntesis , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/farmacología , Conejos , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/uso terapéutico , Saccharomyces cerevisiae , Células Tumorales Cultivadas
20.
Ann N Y Acad Sci ; 886: 58-66, 1999.
Artículo en Inglés | MEDLINE | ID: mdl-10667203

RESUMEN

The growth of solid tumors has been shown to depend on neovascularization. By understanding the mechanisms that control the neovascular response, it may be possible to design therapeutic strategies to selectively prevent or halt pathologic vascular growth and restrain cancer progression. Thrombospondin-1 is an extracellular matrix protein that among several functions suppresses capillary growth in angiogenesis assays. We have demonstrated that within the context of the mammary gland TSP1 can modulate normal development of blood vessels. Expression of TSP1 in transgenic animals under the control of the MMTV promoter was associated with a 50-72% reduction in capillary growth. In addition, TSP1 reduced tumor size in transgenic overexpressors. The data suggest an important role for TSP1 in modulating vascular growth in both normal and pathologic tissues. The antiangiogenic region of TSP1 has been mapped to the type I (properdin) repeats. To identify novel proteins with such a domain, we have cloned two cDNAs (METH-1 and METH-2) which also have antiangiogenic properties. In addition to carboxyterminal thrombospondin-like domains they also contain metalloproteinase and disintegrin sequences. Expression of both proteins is broad but nonoverlapping. Recombinant fragments from these sequences have strong antiangiogenic potential in the CAM and cornea pocket assays. At the same molar ratio, METH-1 and METH-2 are about 20-fold more potent than TSP1. We predict that these proteins are likely endogenous modulators of vascular growth with relevant therapeutic potential in cancer and other disease states.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Metaloproteínas/farmacología , Trombospondinas/farmacología , Secuencia de Aminoácidos , Inhibidores de la Angiogénesis/química , Animales , Animales Modificados Genéticamente , Metaloproteínas/química , Datos de Secuencia Molecular , Neoplasias Experimentales/irrigación sanguínea , Neoplasias Experimentales/patología , Homología de Secuencia de Aminoácido , Trombospondinas/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA